Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 23(8): 101356, 2020 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-32731169

RESUMEN

TLR3, a major innate immune pattern recognition receptor of RNA viruses, triggers inflammatory response through the transcription factor NF-κB. However, a genome-wide understanding of the genes and mechanisms regulating TLR3-mediated NF-κB activation is incomplete. We herein report the results of a human genome-wide RNAi screen that identified 591 proteins regulating TLR3-mediated NF-κB response. Bioinformatics analysis revealed several signaling modules including linear ubiquitination assembly complex and mediator protein complex network as regulators of TLR3 signaling. We further characterized the kinase ATM as a previously unknown positive regulator of TLR3 signaling. TLR3 pathway stimulation induced ATM phosphorylation and promoted interaction of ATM with TAK1, NEMO, IKKα, and IKKß. Furthermore, ATM was determined to coordinate the assembly of NEMO with TAK1, IKKα, and IKKß during TLR3 signaling. This study provided a comprehensive understanding of TLR3-mediated inflammatory signaling regulation and established a role for ATM in innate immune response.

2.
Biomolecules ; 7(3)2017 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-28758979

RESUMEN

In vitro transcribed (IVT) mRNA is increasingly applied in lieu of DNA to deliver reprogramming genes to fibroblasts for stem cell derivation. However, IVT mRNA induces interferon (IFN) responses from mammalian cells that reduces transfection efficiency. It has been previously suggested that small molecule inhibitors of IFN are a viable strategy to enhance mRNA transfection efficiency. Herein, we screen a list of commercially available small molecules, including published IFN inhibitors, for their potential to enhance mRNA transfection in BJ fibroblasts. Transfection enhancement is quantified by relative mean fluorescence intensity of translated green fluorescent protein (GFP) in treated cells compared to dimethyl sulfoxide treated controls. Within toxicological constrains, all tested small molecules did not enhance mRNA transfection in BJ fibroblasts while a third of the tested compounds unexpectedly inhibited GFP expression even though IFN-ß production is inhibited. Based on the results of our study, we conclude that small molecule inhibitors, including IFN inhibitors, tested in this study do not enhance in vitro mRNA transfection efficiency in human fibroblasts.


Asunto(s)
Interferones/antagonistas & inhibidores , ARN Mensajero/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Transfección , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dimetilsulfóxido/farmacología , Evaluación Preclínica de Medicamentos , Fibroblastos/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Nanopartículas , Transcripción Genética
3.
Mol Cell Biol ; 37(6)2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-27956705

RESUMEN

Transcription of type I interferon genes during RNA virus infection requires signal communication between several pattern recognition receptor (PRR)-adaptor complexes located at distinct subcellular membranous compartments and a central cytoplasmic TBK1-interferon regulatory factor 3 (IRF3) kinase-transcription factor module. However, how the cell integrates signal transduction through spatially distinct modules of antiviral signaling pathways is less defined. RIG-I is a major cytosolic PRR involved in the control of several RNA viruses. Here we identify ArfGAP domain-containing protein 2 (ADAP2) as a key novel scaffolding protein that integrates different modules of the RIG-I pathway, located at distinct subcellular locations, and mediates cellular antiviral type I interferon production. ADAP2 served to bridge the mitochondrial membrane-bound upstream RIG-I adaptor MAVS and the downstream cytosolic complex of NEMO (regulatory subunit of TBK1), TBK1, and IRF3, leading to IRF3 phosphorylation. Furthermore, independently, ADAP2 also functioned as a major orchestrator of the interaction of TBK1 with NEMO and IRF3. Mutational and in vitro cell-free reconstituted RIG-I signaling assay-based analyses identified that the ArfGAP domain of ADAP2 mediates the interferon response. TRAF3 acted as a trigger for ADAP2 to recruit RIG-I pathway component proteins into a single macromolecular complex. This study provides important novel insights into the assembly and integration of different modules of antiviral signaling cascades.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/metabolismo , Interferón Tipo I/biosíntesis , Transducción de Señal , Sistema Libre de Células , Proteínas Activadoras de GTPasa/genética , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Modelos Biológicos , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Receptores Inmunológicos , Receptores de Reconocimiento de Patrones/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Transcripción Genética/efectos de los fármacos , Vesiculovirus/fisiología
4.
Sci Rep ; 6: 36179, 2016 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-27782195

RESUMEN

Type I interferon (IFN-I) mediated innate immune response controls virus infections by inducing the expression of interferon stimulated genes (ISGs). Although ubiquitination plays key roles in immune signaling regulation, a human genome-wide understanding of the role of E3 ubiquitin ligases in interferon mediated ISG induction is lacking. Here, we report a genome-wide profiling of the effect of ectopic expression of 521 E3 ubiquitin ligases and substrate recognition subunits encoded in the human genome (which constitutes 84.4% of all ubiquitination related genes encoded in the human genome, hereafter termed Human Ubiquitome) on IFNß mediated induction of interferon stimulated DNA response element (ISRE) driven reporter activity. We identified 96 and 42 genes of the human ubiquitome as novel negative and positive regulators of interferon signaling respectively. Furthermore, we characterized DCST1 as a novel E3 ubiquitin ligase negatively regulating interferon response. Ectopic expression and gene silencing of DCST1 respectively attenuated and increased ISRE reporter activity. DCST1 regulated Type I interferon signaling by interacting with and promoting ubiquitination-mediated degradation of STAT2, an essential component of antiviral gene induction. In summary, this study provided a systems level view on the role of human ubiquitination associated genes in Type I interferon response.


Asunto(s)
Interferón Tipo I/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Ubiquitinadas/metabolismo , Biología Computacional , Genes Reporteros , Células HEK293 , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón beta/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal , Transcripción Genética/efectos de los fármacos , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Proteínas Ubiquitinadas/genética
5.
Methods Mol Biol ; 1435: 115-27, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27188554

RESUMEN

The West Nile virus (WNV) infection is a major medical problem for humans and some domesticated animals. WNV infection of host cells involves the interplay of the virus with several host factors. Identification of the host factors impacting on WNV infection can enhance our understanding of virus infection mechanisms, host immune defense mechanisms, and also reveal novel host targets that can be developed as antivirals. RNA interference (RNAi) is a highly efficient genetic tool to discover host genes involved in WNV infection at a genome scale. Here, we describe a protocol for conducting human genome wide RNAi screen to discover novel host factors associated with WNV infection of human cells.


Asunto(s)
Tratamiento con ARN de Interferencia/métodos , Fiebre del Nilo Occidental/genética , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/fisiología , Evaluación Preclínica de Medicamentos , Regulación Viral de la Expresión Génica , Genoma Humano , Células HeLa , Humanos , Transfección , Replicación Viral , Fiebre del Nilo Occidental/terapia , Virus del Nilo Occidental/genética
6.
Int J Mol Sci ; 17(3): 291, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26927085

RESUMEN

The recent emergence of multiple technologies for modifying gene structure has revolutionized mammalian biomedical research and enhanced the promises of gene therapy. Over the past decade, RNA interference (RNAi) based technologies widely dominated various research applications involving experimental modulation of gene expression at the post-transcriptional level. Recently, a new gene editing technology, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and the CRISPR-associated protein 9 (Cas9) (CRISPR/Cas9) system, has received unprecedented acceptance in the scientific community for a variety of genetic applications. Unlike RNAi, the CRISPR/Cas9 system is bestowed with the ability to introduce heritable precision insertions and deletions in the eukaryotic genome. The combination of popularity and superior capabilities of CRISPR/Cas9 system raises the possibility that this technology may occupy the roles currently served by RNAi and may even make RNAi obsolete. We performed a comparative analysis of the technical aspects and applications of the CRISPR/Cas9 system and RNAi in mammalian systems, with the purpose of charting out a predictive picture on whether the CRISPR/Cas9 system will eclipse the existence and future of RNAi. The conclusion drawn from this analysis is that RNAi will still occupy specific domains of biomedical research and clinical applications, under the current state of development of these technologies. However, further improvements in CRISPR/Cas9 based technology may ultimately enable it to dominate RNAi in the long term.


Asunto(s)
Sistemas CRISPR-Cas , Marcación de Gen/métodos , Interferencia de ARN , ARN Interferente Pequeño/genética , Animales , Marcación de Gen/tendencias , Humanos
7.
Toxicol Lett ; 238(3): 20-9, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26253280

RESUMEN

Although building constructions are a recurring part of urbanization, the health risk of particulate matters (PM) originating from such activities have seldom been subjected to detailed studies. We sought to characterize the relative risk of air borne PM collected from different heights (ground and top floor) of a building adjacent to a building under early phase of construction. We determined the physico-chemical properties such as size and shape, elemental composition and surface charge of the PM. The oxidative stress dependent cytotoxic and pro-inflammatory responses were assessed in BEAS-2B and RAW 264.7 cell lines using high-content-screening platforms. In comparison to top floor, the total mass of PM collected from ground floor was two-three folds higher and the mass fraction was dominated by PM20-35. Elemental analysis showed abundance of Si, Al, K, Ca and Fe in bigger PM while for PM0.25-0.5 it was mostly constituted by C and crystals rich in S and K. PM caused NFκB activation, secretion of pro-inflammatory cytokines and cytotoxicity wherein PM0.25-0.5 was the most potent among the tested PM. Estimated exposure level and lung burden together with the data on hazard potential were used for developing a MATLAB based risk-assessment model which suggested that the potential for health risk is relatively higher at the ground floor. Our studies demonstrated differences in, relative abundance of PM, their physicochemical and biological properties collected from different heights adjacent to a construction site and showed that relative health risk is higher at the ground floor.


Asunto(s)
Contaminantes Atmosféricos/química , Contaminantes Atmosféricos/toxicidad , Contaminación del Aire Interior/análisis , Monitoreo del Ambiente , Tamaño de la Partícula , Material Particulado/toxicidad , Animales , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Macrófagos/efectos de los fármacos , Ratones , Microscopía Electrónica de Rastreo , FN-kappa B/genética , FN-kappa B/metabolismo , Medición de Riesgo
8.
BMC Cell Biol ; 16: 17, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26084399

RESUMEN

BACKGROUND: Cells contain several inositol pyrophosphates (PP-InsPs; also known as diphosphoinositol polyphosphates), which play pivotal roles in cellular and organismic homeostasis. It has been proposed that determining mechanisms of compartmentation of the synthesis of a particular PP-InsP is key to understanding how each of them may exert a specific function. Human PPIP5K2 (hPPIP5K2), one of the key enzymes that synthesizes PP-InsPs, contains a putative consensus sequence for a nuclear localization signal (NLS). However, such in silico analysis has limited predictive power, and may be complicated by phosphorylation events that can dynamically modulate NLS function. We investigated if this candidate NLS is functional and regulated, using the techniques of cell biology, mutagenesis and mass spectrometry. RESULTS: Multiple sequence alignments revealed that the metazoan PPIP5K2 family contains a candidate NLS within a strikingly well-conserved 63 amino-acid domain. By analyzing the distribution of hPPIP5K2-GFP in HEK293T cells with the techniques of confocal microscopy and imaging flow cytometry, we found that a distinct pool of hPPIP5K2 is present in the nucleus. Imaging flow cytometry yielded particular insight into the characteristics of the nuclear hPPIP5K2 sub-pool, through a high-throughput, statistically-robust analysis of many hundreds of cells. Mutagenic disruption of the candidate NLS in hPPIP5K2 reduced its degree of nuclear localization. Proximal to the NLS is a Ser residue (S1006) that mass spectrometry data indicate is phosphorylated inside cells. The degree of nuclear localization of hPPIP5K2 was increased when S1006 was rendered non-phosphorylatable by its mutation to Ala. Conversely, a S1006D phosphomimetic mutant of hPPIP5K2 exhibited a lower degree of nuclear localization. CONCLUSIONS: The current study describes for the first time the functional significance of an NLS in the conserved PPIP5K2 family. We have further demonstrated that there is phosphorylation of a Ser residue that is proximal to the NLS of hPPIP5K2. These conclusions draw attention to nuclear compartmentation of PPIP5K2 as being a physiologically relevant and covalently-regulated event. Our study also increases general insight into the consensus sequences of other NLSs, the functions of which might be similarly regulated.


Asunto(s)
Núcleo Celular/metabolismo , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Fosfopéptidos/análisis , Fosforilación , Fosfotransferasas (Aceptor del Grupo Fosfato)/química , Fosfotransferasas (Aceptor del Grupo Fosfato)/genética , Transporte de Proteínas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Alineación de Secuencia , Espectrometría de Masas en Tándem
9.
PLoS Pathog ; 10(2): e1003981, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586175

RESUMEN

The pattern recognition receptor RIG-I is critical for Type-I interferon production. However, the global regulation of RIG-I signaling is only partially understood. Using a human genome-wide RNAi-screen, we identified 226 novel regulatory proteins of RIG-I mediated interferon-ß production. Furthermore, the screen identified a metabolic pathway that synthesizes the inositol pyrophosphate 1-IP7 as a previously unrecognized positive regulator of interferon production. Detailed genetic and biochemical experiments demonstrated that the kinase activities of IPPK, PPIP5K1 and PPIP5K2 (which convert IP5 to1-IP7) were critical for both interferon induction, and the control of cellular infection by Sendai and influenza A viruses. Conversely, ectopically expressed inositol pyrophosphate-hydrolases DIPPs attenuated interferon transcription. Mechanistic experiments in intact cells revealed that the expression of IPPK, PPIP5K1 and PPIP5K2 was needed for the phosphorylation and activation of IRF3, a transcription factor for interferon. The addition of purified individual inositol pyrophosphates to a cell free reconstituted RIG-I signaling assay further identified 1-IP7 as an essential component required for IRF3 activation. The inositol pyrophosphate may act by ß-phosphoryl transfer, since its action was not recapitulated by a synthetic phosphonoacetate analogue of 1-IP7. This study thus identified several novel regulators of RIG-I, and a new role for inositol pyrophosphates in augmenting innate immune responses to viral infection that may have therapeutic applications.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interferón Tipo I/inmunología , Monoéster Fosfórico Hidrolasas/inmunología , Receptores de Ácido Retinoico/inmunología , Transducción de Señal/inmunología , Humanos , Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/inmunología , ARN Interferente Pequeño
10.
Viruses ; 6(2): 683-708, 2014 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-24517970

RESUMEN

West Nile (WNV) and Dengue (DENV) viruses are major arboviral human pathogens belonging to the genus Flavivirus. At the current time, there are no approved prophylactics (e.g., vaccines) or specific therapeutics available to prevent or treat human infections by these pathogens. Due to their minimal genome, these viruses require many host molecules for their replication and this offers a therapeutic avenue wherein host factors can be exploited as treatment targets. Since several host factors appear to be shared by many flaviviruses the strategy may result in pan-flaviviral inhibitors and may also attenuate the rapid emergence of drug resistant mutant viruses. The scope of this strategy is greatly enhanced by the recent en masse identification of host factors impacting on WNV and DENV infection. Excellent proof-of-principle experimental demonstrations for host-targeted control of infection and infection-induced pathogenesis have been reported for both WNV and DENV. These include exploiting not only those host factors supporting infection, but also targeting host processes contributing to pathogenesis and innate immune responses. While these early studies validated the host-targeting approach, extensive future investigations spanning a range of aspects are needed for a successful deployment in humans.


Asunto(s)
Antivirales/aislamiento & purificación , Virus del Dengue/fisiología , Dengue/tratamiento farmacológico , Dengue/terapia , Interacciones Huésped-Patógeno , Fiebre del Nilo Occidental/tratamiento farmacológico , Virus del Nilo Occidental/fisiología , Animales , Descubrimiento de Drogas/tendencias , Humanos
11.
Proc Natl Acad Sci U S A ; 111(7): 2722-7, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24550301

RESUMEN

Viruses must evade the host innate defenses for replication and dengue is no exception. During secondary infection with a heterologous dengue virus (DENV) serotype, DENV is opsonized with sub- or nonneutralizing antibodies that enhance infection of monocytes, macrophages, and dendritic cells via the Fc-gamma receptor (FcγR), a process termed antibody-dependent enhancement of DENV infection. However, this enhancement of DENV infection is curious as cross-linking of activating FcγRs signals an early antiviral response by inducing the type-I IFN-stimulated genes (ISGs). Entry through activating FcγR would thus place DENV in an intracellular environment unfavorable for enhanced replication. Here we demonstrate that, to escape this antiviral response, antibody-opsonized DENV coligates leukocyte Ig-like receptor-B1 (LILRB1) to inhibit FcγR signaling for ISG expression. This immunoreceptor tyrosine-based inhibition motif-bearing receptor recruits Src homology phosphatase-1 to dephosphorylate spleen tyrosine kinase (Syk). As Syk is a key intermediate of FcγR signaling, LILRB1 coligation resulted in reduced ISG expression for enhanced DENV replication. Our findings suggest a unique mechanism for DENV to evade an early antiviral response for enhanced infection.


Asunto(s)
Acrecentamiento Dependiente de Anticuerpo/fisiología , Antígenos CD/metabolismo , Virus del Dengue/metabolismo , Dengue/fisiopatología , Receptores Inmunológicos/metabolismo , Acrecentamiento Dependiente de Anticuerpo/inmunología , Western Blotting , Línea Celular , Dengue/inmunología , Virus del Dengue/fisiología , Humanos , Receptor Leucocitario Tipo Inmunoglobulina B1 , Análisis por Micromatrices , ARN Interferente Pequeño/genética , Receptores de IgG/metabolismo
12.
PLoS Pathog ; 7(9): e1002189, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21909258

RESUMEN

West Nile (WNV), dengue (DENV) and yellow fever (YFV) viruses are (re)emerging, mosquito-borne flaviviruses that cause human disease and mortality worldwide. Alterations in mosquito gene expression common and unique to individual flaviviral infections are poorly understood. Here, we present a microarray analysis of the Aedes aegypti transcriptome over time during infection with DENV, WNV or YFV. We identified 203 mosquito genes that were ≥ 5-fold differentially up-regulated (DUR) and 202 genes that were ≥ 10-fold differentially down-regulated (DDR) during infection with one of the three flaviviruses. Comparative analysis revealed that the expression profile of 20 DUR genes and 15 DDR genes was quite similar between the three flaviviruses on D1 of infection, indicating a potentially conserved transcriptomic signature of flaviviral infection. Bioinformatics analysis revealed changes in expression of genes from diverse cellular processes, including ion binding, transport, metabolic processes and peptidase activity. We also demonstrate that virally-regulated gene expression is tissue-specific. The overexpression of several virally down-regulated genes decreased WNV infection in mosquito cells and Aedes aegypti mosquitoes. Among these, a pupal cuticle protein was shown to bind WNV envelope protein, leading to inhibition of infection in vitro and the prevention of lethal WNV encephalitis in mice. This work provides an extensive list of targets for controlling flaviviral infection in mosquitoes that may also be used to develop broad preventative and therapeutic measures for multiple flaviviruses.


Asunto(s)
Aedes/virología , Virus del Dengue/fisiología , Transcriptoma/genética , Virus del Nilo Occidental/fisiología , Virus de la Fiebre Amarilla/fisiología , Aedes/genética , Animales , Línea Celular , Virus del Dengue/genética , Regulación hacia Abajo , Femenino , Infecciones por Flavivirus/genética , Infecciones por Flavivirus/prevención & control , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica , Proteínas de Insectos/biosíntesis , Ratones , Ratones Endogámicos C57BL , Virus del Nilo Occidental/genética , Virus de la Fiebre Amarilla/genética
13.
Virology ; 417(1): 179-87, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21700306

RESUMEN

West Nile and dengue viruses are (re)emerging mosquito-borne flaviviruses that cause significant morbidity and mortality in man. The identification of mosquito proteins that associate with flaviviruses may provide novel targets to inhibit infection of the vector or block transmission to humans. Here, a tandem affinity purification (TAP) assay was used to identify 18 mosquito proteins that interact with dengue and West Nile capsid, envelope, NS2A or NS2B proteins. We further analyzed the interaction of mosquito cadherin with dengue and West Nile virus envelope protein using co-immunoprecipitation and immunofluorescence. Blocking the function of select mosquito factors, including actin, myosin, PI3-kinase and myosin light chain kinase, reduced both dengue and West Nile virus infection in mosquito cells. We show that the TAP method may be used in insect cells to accurately identify flaviviral-host protein interactions. Our data also provides several targets for interrupting flavivirus infection in mosquito vectors.


Asunto(s)
Culicidae/virología , Virus del Dengue/metabolismo , Proteínas de Insectos/metabolismo , Insectos Vectores/virología , Proteínas Virales/metabolismo , Virus del Nilo Occidental/metabolismo , Animales , Cadherinas/metabolismo , Culicidae/metabolismo , Insectos Vectores/metabolismo , Unión Proteica
14.
Cell ; 142(5): 714-25, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20797779

RESUMEN

West Nile virus (WNV) is the most common arthropod-borne flavivirus in the United States; however, the vector ligand(s) that participate in infection are not known. We now show that an Aedes aegypti C-type lectin, mosGCTL-1, is induced by WNV, interacts with WNV in a calcium-dependent manner, and facilitates infection in vivo and in vitro. A mosquito homolog of human CD45 in A. aegypti, designated mosPTP-1, recruits mosGCTL-1 to enable viral attachment to cells and to enhance viral entry. In vivo experiments show that mosGCTL-1 and mosPTP-1 function as part of the same pathway and are critical for WNV infection of mosquitoes. A similar phenomenon was also observed in Culex quinquefasciatus, a natural vector of WNV, further demonstrating that these genes participate in WNV infection. During the mosquito blood-feeding process, WNV infection was blocked in vivo with mosGCTL-1 antibodies. A molecular understanding of flaviviral-arthropod interactions may lead to strategies to control viral dissemination in nature.


Asunto(s)
Aedes/virología , Culex/virología , Proteínas de Insectos/metabolismo , Lectinas Tipo C/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Internalización del Virus , Virus del Nilo Occidental/fisiología , Animales , Humanos , Antígenos Comunes de Leucocito/química
15.
J Immunol ; 183(1): 650-60, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19535627

RESUMEN

West Nile virus is an emerging pathogen that can cause fatal neurological disease. A recombinant human mAb, mAb11, has been described as a candidate for the prevention and treatment of West Nile disease. Using a yeast surface display epitope mapping assay and neutralization escape mutant, we show that mAb11 recognizes the fusion loop, at the distal end of domain II of the West Nile virus envelope protein. Ab mAb11 cross-reacts with all four dengue viruses and provides protection against dengue (serotypes 2 and 4) viruses. In contrast to the parental West Nile virus, a neutralization escape variant failed to cause lethal encephalitis (at higher infectious doses) or induce the inflammatory responses associated with blood-brain barrier permeability in mice, suggesting an important role for the fusion loop in viral pathogenesis. Our data demonstrate that an intact West Nile virus fusion loop is critical for virulence, and that human mAb11 targeting this region is efficacious against West Nile virus infection. These experiments define the molecular determinant on the envelope protein recognized by mAb11 and demonstrate the importance of this region in causing West Nile encephalitis.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Sitios de Unión de Anticuerpos , Péptidos/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales de Fusión/inmunología , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/patogenicidad , Animales , Anticuerpos Monoclonales/uso terapéutico , Línea Celular , Reacciones Cruzadas , Virus del Dengue/inmunología , Virus del Dengue/patogenicidad , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Péptidos/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/metabolismo , Fiebre del Nilo Occidental/terapia , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/inmunología
16.
Antiviral Res ; 82(3): 166-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19135091

RESUMEN

West Nile virus (WNV) is an emerging human pathogen for which specific antiviral therapy has not been developed. The therapeutic potential of RNA interference (RNAi) as a sequence-specific inhibitor of WNV has been well demonstrated. Although multiple siRNA targets have been identified within the genomic coding region, targets within the untranslated regions (UTR), which encode cis-acting regulatory elements, remain relatively unknown. In WNV and other flaviviruses, UTRs are located at the genomic termini. These regions form complex secondary structures, which pose difficulty when designing effective siRNA targets. In this study, we report the identification of siRNA targets in the WNV 3' UTR. These targets were selected by siRNA predictor algorithms, and synthesized as short hairpin RNA sequences from a plasmid-based expression system. Vero cells stably expressing these sequences had greatly diminished ability to support WNV replication but not the related dengue virus, demonstrating that the siRNAs were effective and suppressed WNV viral replication in a sequence-specific manner. The siRNAs developed in this study could function as potential antiviral therapeutics and as genetic tools to investigate the role of 3' UTR in WNV pathogenesis.


Asunto(s)
Regiones no Traducidas 3' , Antivirales/farmacología , ARN Interferente Pequeño/genética , ARN Viral/genética , Replicación Viral/efectos de los fármacos , Virus del Nilo Occidental/genética , Animales , Secuencia de Bases , Chlorocebus aethiops , Virus del Dengue/genética , Virus del Dengue/fisiología , Humanos , Datos de Secuencia Molecular , ARN Interferente Pequeño/farmacología , Células Vero , Ensayo de Placa Viral , Virus del Nilo Occidental/fisiología
17.
Cell ; 139(7): 1243-54, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20064371

RESUMEN

Influenza viruses exploit host cell machinery to replicate, resulting in epidemics of respiratory illness. In turn, the host expresses antiviral restriction factors to defend against infection. To find host cell modifiers of influenza A H1N1 viral infection, we used a functional genomic screen and identified over 120 influenza A virus-dependency factors with roles in endosomal acidification, vesicular trafficking, mitochondrial metabolism, and RNA splicing. We discovered that the interferon-inducible transmembrane proteins IFITM1, 2, and 3 restrict an early step in influenza A viral replication. The IFITM proteins confer basal resistance to influenza A virus but are also inducible by interferons type I and II and are critical for interferon's virustatic actions. Further characterization revealed that the IFITM proteins inhibit the early replication of flaviviruses, including dengue virus and West Nile virus. Collectively this work identifies a family of antiviral restriction factors that mediate cellular innate immunity to at least three major human pathogens.


Asunto(s)
Infecciones por Flavivirus/inmunología , Gripe Humana/inmunología , Proteínas de la Membrana/inmunología , Animales , Antígenos de Diferenciación , Línea Celular Tumoral , Virus del Dengue/inmunología , Humanos , Inmunidad Innata , Virus de la Influenza A/inmunología , Interferones/inmunología , Ratones , Proteínas de Unión al ARN/inmunología , Virus del Nilo Occidental/inmunología , Virus del Nilo Occidental/fisiología
18.
Nature ; 455(7210): 242-5, 2008 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-18690214

RESUMEN

West Nile virus (WNV), and related flaviviruses such as tick-borne encephalitis, Japanese encephalitis, yellow fever and dengue viruses, constitute a significant global human health problem. However, our understanding of the molecular interaction of such flaviviruses with mammalian host cells is limited. WNV encodes only 10 proteins, implying that it may use many cellular proteins for infection. WNV enters the cytoplasm through pH-dependent endocytosis, undergoes cycles of translation and replication, assembles progeny virions in association with endoplasmic reticulum, and exits along the secretory pathway. RNA interference (RNAi) presents a powerful forward genetics approach to dissect virus-host cell interactions. Here we report the identification of 305 host proteins that affect WNV infection, using a human-genome-wide RNAi screen. Functional clustering of the genes revealed a complex dependence of this virus on host cell physiology, requiring a wide variety of molecules and cellular pathways for successful infection. We further demonstrate a requirement for the ubiquitin ligase CBLL1 in WNV internalization, a post-entry role for the endoplasmic-reticulum-associated degradation pathway in viral infection, and the monocarboxylic acid transporter MCT4 as a viral replication resistance factor. By extending this study to dengue virus, we show that flaviviruses have both overlapping and unique interaction strategies with host cells. This study provides a comprehensive molecular portrait of WNV-human cell interactions that forms a model for understanding single plus-stranded RNA virus infection, and reveals potential antiviral targets.


Asunto(s)
Interferencia de ARN , Fiebre del Nilo Occidental/genética , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/fisiología , Biología Computacional , Virus del Dengue/fisiología , Retículo Endoplásmico/metabolismo , Perfilación de la Expresión Génica , Genoma Humano , VIH , Células HeLa , Humanos , Inmunidad/genética , Transportadores de Ácidos Monocarboxílicos/deficiencia , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Unión Proteica , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/genética , Vesiculovirus , Replicación Viral
19.
Biochemistry ; 47(19): 5354-67, 2008 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-18419132

RESUMEN

The selectivity filter of most K+ channels contains a highly conserved Thr residue that uniquely forms the S4 binding site for K+ by dual coordination with the backbone carbonyl oxygen and side chain hydroxyl of the same residue. This study examines the effect of mutations of Thr75 in the S4 site of theKcsA K+ channel on the cation dependence of the thermal stability of the tetramer, a phenomenon that reflects the structural role of cations in the filter. Conservative mutations of Thr75 destabilize the tetramer and alter its temperature dependence. Replacement of Thr with Ala or Cys lowers the apparent affinity ofK+, Rb+, and Cs+ for tetramer stabilization by factors ranging from 4- to 14-fold. These same mutations lower the apparent affinity of Ba2+ by approximately 10(3)- or approximately 10(4)-fold for Ala and Cys substitution, respectively,consistent with the known preference of the S4 site for Ba2+. In contrast, substitution of Ala or Cys at T75 anomalously enhances the ability of Na+ to stabilize the tetramer, suggesting that the native Thr residue at S4 is important for ultrahigh K+/Na+ selectivity of K+ channel pores. Elevated temperature orCu2+ cation catalyzes formation of covalent dimers of the T75C mutant of KcsA via formation of disulfide bonds between Cys residues of adjacent subunits. Thiophilic cations such as Hg2+ and Ag+ specifically protect the T75C tetramer against heat-induced dimer formation, demonstrating the contribution of cation interactions to tetramer stability in a channel with a non-native S4 site engineered to bind foreign cations.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Secuencia Conservada , Canales de Potasio/química , Canales de Potasio/metabolismo , Streptomyces lividans/química , Temperatura , Treonina/metabolismo , Proteínas Bacterianas/genética , Cationes/química , Dimerización , Mutación/genética , Canales de Potasio/genética , Unión Proteica , Desnaturalización Proteica , Sensibilidad y Especificidad , Compuestos de Sulfhidrilo/química , Treonina/genética
20.
J Virol ; 81(9): 4881-5, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17301152

RESUMEN

The mechanisms of cellular entry of dengue and West Nile viruses are not well characterized. We show that both these viruses enter HeLa cells by clathrin-dependent endocytosis and require vacuolar acidic pH. Inhibition of the GTPase Rab 5 or 7, which regulates transport to early or late endosomes, respectively, demonstrated that Rab 5 was essential for survival of both dengue and West Nile virus. These data broaden our understanding of the pathways required for productive dengue and West Nile virus infection and may facilitate new strategies for combating disease.


Asunto(s)
Virus del Dengue/metabolismo , Internalización del Virus , Virus del Nilo Occidental/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Endocitosis/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Interferencia de ARN , Vacuolas/química , Vacuolas/metabolismo , Proteínas de Unión al GTP rab5/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...